1
|
Hundhausen N, Majumder S, Xiao Y, Haeusl SS, Goehler H, Seal R, Chiarolla CM, Rosenwald A, Eyrich M, Cicin-Sain L, Berberich-Siebelt F. NFAT single-deficient murine T cells reduce the risk of aGvHD while controlling cytomegalovirus infection. iScience 2025; 28:111937. [PMID: 40028277 PMCID: PMC11872454 DOI: 10.1016/j.isci.2025.111937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/13/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
NFAT is a family of transcription factors whose activation is inhibited by calcineurin inhibitors (CNIs). In allogeneic hematopoietic stem cell transplantation (allo-HCT), CNIs are employed to prevent and treat graft-versus-host disease (GvHD). Unfortunately, control of cytomegalovirus (CMV), which exacerbates clinical outcomes, is simultaneously lost. Since single NFAT deficiency in T cells ameliorates GvHD in our major mismatch model, we investigated whether protection is maintained during CMV infection. Reassuringly, NFAT-deficient T cells still improved GvHD upon acute CMV infection and after allo-HCT in latently CMV-infected mice, showing reduced proinflammatory and cytotoxic potential. In sharp contrast, CMV-specific NFAT-deficient CD8+ inflated memory T cells expanded more and with higher levels of interferon gamma (IFN-γ) and GzmB expression, effectively controlling CMV. Notably, NFAT-deficient inflated memory T cells could migrate to non-lymphoid tissues and fight CMV. Therefore, CMV infection does not interfere with the protective effect of NFAT inhibition to attenuate GvHD while allowing an anti-CMV response.
Collapse
Affiliation(s)
- Nadine Hundhausen
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Snigdha Majumder
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Yin Xiao
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Sigrun S. Haeusl
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Helen Goehler
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Rishav Seal
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | | | - Andreas Rosenwald
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
- Comprehensive Cancer Centre Mainfranken, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Matthias Eyrich
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Luka Cicin-Sain
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine, a Joint Venture of Helmholtz Centre for Infection Research and Medical School Hannover, Hannover, Germany
| | | |
Collapse
|
2
|
van Pul L, Stunnenberg M, Kroeze S, van Dort KA, Boeser-Nunnink BDM, Harskamp AM, Geijtenbeek TBH, Kootstra NA. Energy demanding RNA and protein metabolism drive dysfunctionality of HIV-specific T cell changes during chronic HIV infection. PLoS One 2024; 19:e0298472. [PMID: 39356699 PMCID: PMC11446443 DOI: 10.1371/journal.pone.0298472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 08/08/2024] [Indexed: 10/04/2024] Open
Abstract
Antiretroviral treatment of HIV infected individuals cannot eliminate the HIV reservoir and immune control of HIV is rarely seen upon treatment interruption. In long-term non-progressors (LTNP), an effective CD8 T cell response is thought to contribute to be immune control of HIV. Here we studied the transcriptional profile of virus specific CD8 T cells during the asymptomatic phase of disease, to gain molecular insights in CD8 T cell functionality in HIV progressors and different groups of LTNP: HLA-B*57 LTNP, non-HLA-B*57 LTNP and individuals carrying the MAVS minor genotype (rs7262903/rs7269320). Principal component analysis revealed distinct overall transcriptional profiles between the groups. The transcription profile of HIV-specific CD8 T cells of LTNP groups was associated with increased cytokine/IL-12 signaling and protein/RNA metabolism pathways, indicating an increased CD8 T cell functionality. Although the transcription profile of CMV-specific CD8 T cells differed from that of HIV-specific CD8 T cells, with mainly an upregulation of gene expression in progressors, similar affected pathways were identified. Moreover, CMV-specific CD8 T cells from progressors showed increased expression of genes related to effector functions and suggests recent antigen exposure. Our data shows that changes in cytokine signaling and the energy demanding RNA and protein metabolism are related to CD8 T cell dysfunction, which may indicate that mitochondrial dysfunction is an important driver of T cell dysfunctionality during chronic HIV infection. Indeed, improvement of mitochondrial function by IL-12 and mitoTempo treatment, enhanced in vitro IFNγ release by PBMC from PWH upon HIV gag and CMV pp65 peptide stimulation. Our study provides new insights into the molecular pathways associated with CD8 T cell mediated immune control of chronic HIV infection which is important for the design of novel treatment strategies to restore or improve the HIV-specific immune response.
Collapse
Affiliation(s)
- Lisa van Pul
- Amsterdam UMC location University of Amsterdam, Laboratory for Viral Immune Pathogenesis, Amsterdam, The Netherlands
- Amsterdam UMC location University of Amsterdam, Department of Experimental Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Melissa Stunnenberg
- Amsterdam UMC location University of Amsterdam, Department of Experimental Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Stefanie Kroeze
- Amsterdam UMC location University of Amsterdam, Laboratory for Viral Immune Pathogenesis, Amsterdam, The Netherlands
- Amsterdam UMC location University of Amsterdam, Department of Experimental Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Karel A van Dort
- Amsterdam UMC location University of Amsterdam, Laboratory for Viral Immune Pathogenesis, Amsterdam, The Netherlands
- Amsterdam UMC location University of Amsterdam, Department of Experimental Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Brigitte D M Boeser-Nunnink
- Amsterdam UMC location University of Amsterdam, Laboratory for Viral Immune Pathogenesis, Amsterdam, The Netherlands
- Amsterdam UMC location University of Amsterdam, Department of Experimental Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Agnes M Harskamp
- Amsterdam UMC location University of Amsterdam, Laboratory for Viral Immune Pathogenesis, Amsterdam, The Netherlands
- Amsterdam UMC location University of Amsterdam, Department of Experimental Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Teunis B H Geijtenbeek
- Amsterdam UMC location University of Amsterdam, Department of Experimental Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Neeltje A Kootstra
- Amsterdam UMC location University of Amsterdam, Laboratory for Viral Immune Pathogenesis, Amsterdam, The Netherlands
- Amsterdam UMC location University of Amsterdam, Department of Experimental Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Han S, Georgiev P, Ringel AE, Sharpe AH, Haigis MC. Age-associated remodeling of T cell immunity and metabolism. Cell Metab 2023; 35:36-55. [PMID: 36473467 PMCID: PMC10799654 DOI: 10.1016/j.cmet.2022.11.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 10/14/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022]
Abstract
Aging results in remodeling of T cell immunity and is associated with poor clinical outcomes in age-related diseases such as cancer. Among the hallmarks of aging, changes in host and cellular metabolism critically affect the development, maintenance, and function of T cells. Although metabolic perturbations impact anti-tumor T cell responses, the link between age-associated metabolic dysfunction and anti-tumor immunity remains unclear. In this review, we summarize recent advances in our understanding of aged T cell metabolism, with a focus on the bioenergetic and immunologic features of T cell subsets unique to the aging process. We also survey insights into mechanisms of metabolic T cell dysfunction in aging and discuss the impacts of aging on the efficacy of cancer immunotherapy. As the average life expectancy continues to increase, understanding the interplay between age-related metabolic reprogramming and maladaptive T cell immunity will be instrumental for the development of therapeutic strategies for older patients.
Collapse
Affiliation(s)
- SeongJun Han
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Peter Georgiev
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Alison E Ringel
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arlene H Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA.
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
4
|
Watanabe N, Mo F, McKenna MK. Impact of Manufacturing Procedures on CAR T Cell Functionality. Front Immunol 2022; 13:876339. [PMID: 35493513 PMCID: PMC9043864 DOI: 10.3389/fimmu.2022.876339] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/15/2022] [Indexed: 12/21/2022] Open
Abstract
The field of chimeric antigen receptor (CAR) modified T cell therapy has rapidly expanded in the past few decades. As of today, there are six CAR T cell products that have been approved by the FDA: KYMRIAH (tisagenlecleucel, CD19 CAR T cells), YESCARTA (axicabtagene ciloleucel, CD19 CAR T cells), TECARTUS (brexucabtagene autoleucel, CD19 CAR T cells), BREYANZI (lisocabtagene maraleucel, CD19 CAR T cells), ABECMA (idecabtagene vicleucel, BCMA CAR T cells) and CARVYKTI (ciltacabtagene autoleucel, BCMA CAR T cells). With this clinical success, CAR T cell therapy has become one of the most promising treatment options to combat cancers. Current research efforts focus on further potentiating its efficacy in non-responding patients and solid tumor settings. To achieve this, recent evidence suggested that, apart from developing next-generation CAR T cells with additional genetic modifications, ex vivo culture conditions could significantly impact CAR T cell functionality - an often overlooked aspect during clinical translation. In this review, we focus on the ex vivo manufacturing process for CAR T cells and discuss how it impacts CAR T cell function.
Collapse
Affiliation(s)
- Norihiro Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | - Feiyan Mo
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Mary Kathryn McKenna
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
5
|
Suàrez-Fernández P, Utrero-Rico A, Sandonis V, García-Ríos E, Arroyo-Sánchez D, Fernández-Ruiz M, Andrés A, Polanco N, González-Cuadrado C, Almendro-Vázquez P, Pérez-Romero P, Aguado JM, Paz-Artal E, Laguna-Goya R. Circulatory follicular helper T lymphocytes associate with lower incidence of CMV infection in kidney transplant recipients. Am J Transplant 2021; 21:3946-3957. [PMID: 34153157 DOI: 10.1111/ajt.16725] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 01/25/2023]
Abstract
Primary infection and/or reactivation of cytomegalovirus (CMV) in kidney transplant recipients (KTR) favor rejection and mortality. T follicular helper cells (TFH) could contribute to protection against CMV. Circulatory TFH (cTFH) were studied pretransplant and early posttransplant in 90 CMV seropositive KTR not receiving antithymocyte globulin or antiviral prophylaxis, followed-up for 1 year. Patients who presented CMV infection had significantly lower cTFH and activated cTFH pretransplant and early posttransplant. Pretransplant activated cTFH were also lower within patients who developed CMV disease. Pre- and 14 days posttransplant activated cTFH were an independent protective factor for CMV infection (HR 0.41, p = .01; and 0.52, p = .02, respectively). KTR with low cTFH 7 days posttransplant (<11.9%) had lower CMV infection-free survival than patients with high cTFH (28.2% vs. 67.6%, p = .002). cTFH were associated with CMV-specific neutralizing antibodies (Nabs). In addition, IL-21 increased interferon-γ secretion by CMV-specific CD8+ T cells in healthy controls. Thus, we show an association between cTFH and lower incidence of CMV infection, probably through their cooperation in CMV-specific Nab production and IL-21-mediated enhancement of CD8+ T cell activity. Moreover, monitoring cTFH pre- and early posttransplant could improve CMV risk stratification and help select KTR catalogued at low/intermediate risk who could benefit from prophylaxis.
Collapse
Affiliation(s)
| | - Alberto Utrero-Rico
- Instituto de Investigación Biomédica Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Virginia Sandonis
- National Center for Microbiology, Instituto de Salud Carlos III, Madrid, Majadahonda, Spain
| | - Estéfani García-Ríos
- National Center for Microbiology, Instituto de Salud Carlos III, Madrid, Majadahonda, Spain
| | - Daniel Arroyo-Sánchez
- Instituto de Investigación Biomédica Hospital 12 de Octubre (imas12), Madrid, Spain.,Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Mario Fernández-Ruiz
- Instituto de Investigación Biomédica Hospital 12 de Octubre (imas12), Madrid, Spain.,Unit of Infectious Diseases, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Amado Andrés
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Natalia Polanco
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | | | - Pilar Pérez-Romero
- National Center for Microbiology, Instituto de Salud Carlos III, Madrid, Majadahonda, Spain
| | - José María Aguado
- Instituto de Investigación Biomédica Hospital 12 de Octubre (imas12), Madrid, Spain.,Unit of Infectious Diseases, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Estela Paz-Artal
- Instituto de Investigación Biomédica Hospital 12 de Octubre (imas12), Madrid, Spain.,Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Department of Immunology, Universidad Complutense de Madrid, Madrid, Spain
| | - Rocío Laguna-Goya
- Instituto de Investigación Biomédica Hospital 12 de Octubre (imas12), Madrid, Spain.,Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
| |
Collapse
|
6
|
Smith CJ, Snyder CM. Inhibitory Molecules PD-1, CD73 and CD39 Are Expressed by CD8 + T Cells in a Tissue-Dependent Manner and Can Inhibit T Cell Responses to Stimulation. Front Immunol 2021; 12:704862. [PMID: 34335618 PMCID: PMC8320728 DOI: 10.3389/fimmu.2021.704862] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/29/2021] [Indexed: 12/16/2022] Open
Abstract
The salivary gland is an important tissue for persistence and transmission of multiple viruses. Previous work showed that salivary gland tissue-resident CD8+ T cells elicited by viruses were poorly functional ex vivo. Using a model of persistent murine cytomegalovirus (MCMV) infection, we now show that CD8+ T cells in the salivary gland and other non-lymphoid tissues of mice express multiple molecules associated with T cell exhaustion including PD-1, CD73 and CD39. Strikingly however, these molecules were expressed independently of virus or antigen. Rather, PD-1-expressing T cells remained PD-1+ after migration into tissues regardless of infection, while CD73 was activated on CD8+ T cells by TGF-β signaling. Blockade of PD-L1, but not CD73, improved cytokine production by salivary gland T cells ex vivo and increased the expression of granzyme B after stimulation within the salivary gland. Nevertheless, salivary-gland localized CD8+ T cells could kill PD-L1-expressing targets in vivo, albeit with modest efficiency, and this was not improved by PD-L1 blockade. Moreover, the impact of PD-L1 blockade on granzyme B expression waned with time. In contrast, the function of kidney-localized T cells was improved by CD73 blockade, but was unaffected by PD-L1 blockade. These data show that tissue localization per se is associated with expression of inhibitory molecules that can impact T cell function, but that the functional impact of this expression is context- and tissue-dependent.
Collapse
Affiliation(s)
- Corinne J Smith
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Christopher M Snyder
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
7
|
Ahmed R, Miners KL, Lahoz-Beneytez J, Jones RE, Roger L, Baboonian C, Zhang Y, Wang ECY, Hellerstein MK, McCune JM, Baird DM, Price DA, Macallan DC, Asquith B, Ladell K. CD57 + Memory T Cells Proliferate In Vivo. Cell Rep 2020; 33:108501. [PMID: 33326780 PMCID: PMC7758161 DOI: 10.1016/j.celrep.2020.108501] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 08/26/2020] [Accepted: 11/18/2020] [Indexed: 01/15/2023] Open
Abstract
A central paradigm in the field of lymphocyte biology asserts that replicatively senescent memory T cells express the carbohydrate epitope CD57. These cells nonetheless accumulate with age and expand numerically in response to persistent antigenic stimulation. Here, we use in vivo deuterium labeling and ex vivo analyses of telomere length, telomerase activity, and intracellular expression of the cell-cycle marker Ki67 to distinguish between two non-exclusive scenarios: (1) CD57+ memory T cells do not proliferate and instead arise via phenotypic transition from the CD57- memory T cell pool; and/or (2) CD57+ memory T cells self-renew via intracompartmental proliferation. Our results provide compelling evidence in favor of the latter scenario and further suggest in conjunction with mathematical modeling that self-renewal is by far the most abundant source of newly generated CD57+ memory T cells. Immunological memory therefore appears to be intrinsically sustainable among highly differentiated subsets of T cells that express CD57.
Collapse
Affiliation(s)
- Raya Ahmed
- Institute for Infection and Immunity, St. George's, University of London, London SW17 0RE, UK
| | - Kelly L Miners
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | | | - Rhiannon E Jones
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Laureline Roger
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Christina Baboonian
- Institute for Infection and Immunity, St. George's, University of London, London SW17 0RE, UK
| | - Yan Zhang
- Institute for Infection and Immunity, St. George's, University of London, London SW17 0RE, UK
| | - Eddie C Y Wang
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Marc K Hellerstein
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| | - Joseph M McCune
- HIV Frontiers Program, Global Health Innovative Technology Solutions, Bill & Melinda Gates Foundation, Seattle, WA 98109, USA
| | - Duncan M Baird
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK; Systems Immunity Research Institute, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK.
| | - Derek C Macallan
- Institute for Infection and Immunity, St. George's, University of London, London SW17 0RE, UK; St George's University Hospitals NHS Foundation Trust, London SW17 0QT, UK.
| | - Becca Asquith
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK.
| | - Kristin Ladell
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK; Neonatal Unit, Singleton Hospital, Swansea Bay University Health Board, Swansea SA2 8QA, UK.
| |
Collapse
|
8
|
Migueles SA, Rogan DC, Gavil NV, Kelly EP, Toulmin SA, Wang LT, Lack J, Ward AJ, Pryal PF, Ludwig AK, Medina RG, Apple BJ, Toumanios CN, Poole AL, Rehm CA, Jones SE, Liang CJ, Connors M. Antigenic Restimulation of Virus-Specific Memory CD8 + T Cells Requires Days of Lytic Protein Accumulation for Maximal Cytotoxic Capacity. J Virol 2020; 94:e01595-20. [PMID: 32907983 PMCID: PMC7654275 DOI: 10.1128/jvi.01595-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 09/05/2020] [Indexed: 02/07/2023] Open
Abstract
In various infections or vaccinations of mice or humans, reports of the persistence and the requirements for restimulation of the cytotoxic mediators granzyme B (GrB) and perforin (PRF) in CD8+ T cells have yielded disparate results. In this study, we examined the kinetics of PRF and GrB mRNA and protein expression after stimulation and associated changes in cytotoxic capacity in virus-specific memory cells in detail. In patients with controlled HIV or cleared respiratory syncytial virus (RSV) or influenza virus infections, all virus-specific CD8+ T cells expressed low PRF levels without restimulation. Following stimulation, they displayed similarly delayed kinetics for lytic protein expression, with significant increases occurring by days 1 to 3 before peaking on days 4 to 6. These increases were strongly correlated with, but were not dependent upon, proliferation. Incremental changes in PRF and GrB percent expression and mean fluorescence intensity (MFI) were highly correlated with increases in HIV-specific cytotoxicity. mRNA levels in HIV-specific CD8+ T-cells exhibited delayed kinetics after stimulation as with protein expression, peaking on day 5. In contrast to GrB, PRF mRNA transcripts were little changed over 5 days of stimulation (94-fold versus 2.8-fold, respectively), consistent with posttranscriptional regulation. Changes in expression of some microRNAs, including miR-17, miR-150, and miR-155, suggested that microRNAs might play a significant role in regulation of PRF expression. Therefore, under conditions of extremely low or absent antigen levels, memory virus-specific CD8+ T cells require prolonged stimulation over days to achieve maximal lytic protein expression and cytotoxic capacity.IMPORTANCE Antigen-specific CD8+ T cells play a major role in controlling most virus infections, primarily by perforin (PRF)- and granzyme B (GrB)-mediated apoptosis. There is considerable controversy regarding whether PRF is constitutively expressed, rapidly increased similarly to a cytokine, or delayed in its expression with more prolonged stimulation in virus-specific memory CD8+ T cells. In this study, the degree of cytotoxic capacity of virus-specific memory CD8+ T cells was directly proportional to the content of lytic molecules, which required antigenic stimulation over several days for maximal levels. This appeared to be modulated by increases in GrB transcription and microRNA-mediated posttranscriptional regulation of PRF expression. Clarifying the requirements for maximal cytotoxic capacity is critical to understanding how viral clearance might be mediated by memory cells and what functions should be induced by vaccines and immunotherapies.
Collapse
Affiliation(s)
- Stephen A Migueles
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniel C Rogan
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Noah V Gavil
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Elizabeth P Kelly
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sushila A Toulmin
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Lawrence T Wang
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Justin Lack
- NIAID Collaborative Bioinformatics Resource (NCBR), Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Maryland, USA
| | - Addison J Ward
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Patrick F Pryal
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Amanda K Ludwig
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Renata G Medina
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Benjamin J Apple
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Christina N Toumanios
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - April L Poole
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Catherine A Rehm
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sara E Jones
- Clinical Research Program Directorate, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Maryland, USA
| | - C Jason Liang
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Mark Connors
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
9
|
Abassi L, Cicin-Sain L. The avid competitors of memory inflation. Curr Opin Virol 2020; 44:162-168. [PMID: 33039898 DOI: 10.1016/j.coviro.2020.08.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/14/2020] [Accepted: 08/16/2020] [Indexed: 12/18/2022]
Abstract
Cytomegaloviruses (CMV) coevolve with their hosts and latently persist in the vast majority of adult mammals. Therefore, persistent T-cell responses to CMV antigens during virus latency offer a fascinating perspective on the evolution of the T-cell repertoire in natural settings. We addressed here the life-long interactions between CMV antigens presented on MHC-I molecules and the CD8 T-cell response. We present the mechanistic evidence from the murine model of CMV infection and put it in context of clinical laboratory results. We will highlight the remarkable parallels in T-cell responses between the two biological systems, and focus in particular on memory inflation as a result of competitive processes, both between viral antigenic peptides and between T-cell receptors on the host's cytotoxic lymphocytes.
Collapse
Affiliation(s)
- Leila Abassi
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Germany
| | - Luka Cicin-Sain
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School (MHH), Germany; Centre for Individualised Infection Medicine (CIIM), A Joint Venture of HZI and MHH, Germany; German Centre for Infection Research (DZIF), Hannover-Braunschweig Site, Germany.
| |
Collapse
|
10
|
Luo XH, Meng Q, Liu Z, Paraschoudi G. Generation of high-affinity CMV-specific T cells for adoptive immunotherapy using IL-2, IL-15, and IL-21. Clin Immunol 2020; 217:108456. [PMID: 32376504 DOI: 10.1016/j.clim.2020.108456] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 12/12/2022]
Abstract
Cytomegalovirus (CMV) infection remains a life-threatening condition in individuals with a suppressed immune system. CMV may also represent a clinically relevant target for immune responses in CMV-positive malignancies. We established a protocol to expand CMV-specific T cells (CMV-T) using peripheral blood mononuclear cells (PBMCs). PBMCs from 16 HLA-A*0201 donors were cultured with a cytokine cocktail comprising IL-2/IL-15/IL-21 along with overlapping peptides from CMV-pp65. Ten days later, T cells were stimulated with anti-CD3 (OKT3) and irradiated autologous PBMCs. CMV-T were detected by HLA-A*0201 CMV-pp65NLVPMVATV wild type and q226a mutant tetramers (for high-affinity T cells), intracellular cytokine staining, a CD107a mobilization assays as well as IFN-γ and TNF-α production in cell culture supernatants. We reliably obtained 50.25 ± 27.27% of CD8+ and 22.08 ± 21.83% of CD4+ T cells post-CMV-pp65 stimulation of PBMCs with a Th1-polarized phenotype and decreased Th2/Th17 responses. Most CD3 + CD8 + tetramer+ T cells were effector-memory cells, particularly among high-affinity CMV-T (q226a CMV-tetramer+). High-affinity CMV-T cells, compared to WT-tetramer+ cells, expressed higher IL-21R and lower FasL post-stimulation with CMV-pp65. The IL-2/IL-15/IL-21 cocktail also promoted CCR6 and CXCR3 expression necessary for T-cell migration into tissues. We have optimized methods for generating high-affinity CMV-specific T cells that can be used for adoptive cellular therapy in clinical practice.
Collapse
Affiliation(s)
- Xiao-Hua Luo
- Therapeutic immunology unit, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Yuzhong District, Chongqing 400016, China.
| | - Qingda Meng
- Therapeutic immunology unit, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Zhenjiang Liu
- Therapeutic immunology unit, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Georgia Paraschoudi
- Therapeutic immunology unit, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
11
|
J. Heath J, D. Grant M. The Immune Response Against Human Cytomegalovirus Links Cellular to Systemic Senescence. Cells 2020; 9:cells9030766. [PMID: 32245117 PMCID: PMC7140628 DOI: 10.3390/cells9030766] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/11/2020] [Accepted: 03/17/2020] [Indexed: 12/12/2022] Open
Abstract
Aging reflects long-term decline in physiological function and integrity. Changes arise at a variable pace governed by time-dependent and -independent mechanisms that are themselves complex, interdependent and variable. Molecular decay produces inferior cells that eventually dominate over healthy counterparts in tissues they comprise. In a form of biological entropy, progression from molecular through cellular to tissue level degeneration culminates in organ disease or dysfunction, affecting systemic health. To better understand time-independent contributors and their potential modulation, common biophysical bases for key molecular and cellular changes underlying age-related physiological deterioration must be delineated. This review addresses the potential contribution of cytomegalovirus (CMV)-driven T cell proliferation to cellular senescence and immunosenescence. We first describe molecular processes imposing cell cycle arrest, the foundation of cellular senescence, then focus on the unique distribution, phenotype and function of CMV-specific CD8+ T cells in the context of cellular senescence and "inflammaging". Their features position CMV infection as a pathogenic accelerant of immune cell proliferation underlying immune senescence. In human immunodeficiency virus (HIV) infection, where increased inflammation and exaggerated anti-CMV immune responses accelerate immune senescence, CMV infection has emerged as a major factor in unhealthy aging. Thus, we speculate on mechanistic links between CMV-specific CD8+ T-cell expansion, immune senescence and prevalence of age-related disorders in HIV infection.
Collapse
Affiliation(s)
- John J. Heath
- Immunology and Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St. John’s, NL A1B 3V6, Canada;
- Lady Davis Institute for Medical Research, Jewish General Hospital, Division of Experimental Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Michael D. Grant
- Immunology and Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St. John’s, NL A1B 3V6, Canada;
- Correspondence:
| |
Collapse
|
12
|
Wilski NA, Snyder CM. From Vaccine Vector to Oncomodulation: Understanding the Complex Interplay between CMV and Cancer. Vaccines (Basel) 2019; 7:E62. [PMID: 31323930 PMCID: PMC6789822 DOI: 10.3390/vaccines7030062] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 12/14/2022] Open
Abstract
Cytomegalovirus (CMV) is a herpesvirus that establishes a persistent, but generally asymptomatic, infection in most people in the world. However, CMV drives and sustains extremely large numbers of antigen-specific T cells and is, therefore, emerging as an exciting platform for vaccines against infectious diseases and cancer. Indeed, pre-clinical data strongly suggest that CMV-based vaccines can sustain protective CD8+ T cell and antibody responses. In the context of vaccines for infectious diseases, substantial pre-clinical studies have elucidated the efficacy and protective mechanisms of CMV-based vaccines, including in non-human primate models of various infections. In the context of cancer vaccines, however, much less is known and only very early studies in mice have been conducted. To develop CMV-based cancer vaccines further, it will be critical to better understand the complex interaction of CMV and cancer. An array of evidence suggests that naturally-acquired human (H)CMV can be detected in cancers, and it has been proposed that HCMV may promote tumor growth. This would obviously be a concern for any therapeutic cancer vaccines. In experimental models, CMV has been shown to play both positive and negative roles in tumor progression, depending on the model studied. However, the mechanisms are still largely unknown. Thus, more studies assessing the interaction of CMV with the tumor microenvironment are needed. This review will summarize the existing literature and major open questions about CMV-based vaccines for cancer, and discuss our hypothesis that the balance between pro-tumor and anti-tumor effects driven by CMV depends on the location and the activity of the virus in the lesion.
Collapse
Affiliation(s)
- Nicole A Wilski
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Christopher M Snyder
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
13
|
The Cytomegalovirus-Specific IL-21 ELISpot Correlates with Allograft Function of Kidney Transplant Recipients. Int J Mol Sci 2018; 19:ijms19123945. [PMID: 30544783 PMCID: PMC6320857 DOI: 10.3390/ijms19123945] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/05/2018] [Accepted: 12/05/2018] [Indexed: 12/17/2022] Open
Abstract
In kidney transplant recipients, the cytomegalovirus (CMV) is frequently causing infection/reactivation and can trigger allograft rejection. To assess the risk of reactivation, the cellular immune response against CMV is increasingly assessed by cellular in vitro methods, such as the interferon (IFN)-γ ELISpot. In the current study we compared the IFN-γ ELISpot with our newly established CMV-specific ELISpot assays determining IL-17A, IL-21, IL-22, granzyme B, and perforin and correlated the results with flow cytometric data and clinical parameters. In 77 kidney transplant recipients, the highest frequency was observed for CMV pp65-specific cells secreting IFN-γ, followed by cells secreting IL-21 (62.9 and 23.2 Δ spot forming cells/105 cells). We observed a positive correlation between the percentage of CMV-specific CD3+ CD4+ CD154+ cells and results of the CMV-specific IL-21 ELISpot (p = 0.002). Results of the CMV pp65-specific IL-21 ELISpot correlated negatively with kidney function (estimated glomerular filtration rate, p = 0.006) and were significantly higher in women (p = 0.005). IL-21, a cytokine involved in aging that is secreted by activated CD4+ T cells, may also impact on allograft function. Thus, the CMV-specific IL-21 ELISpot could become a new tool to assess if CMV seropositivity represents a hazard for the graft.
Collapse
|
14
|
Waters S, Brook E, Lee S, Estiasari R, Ariyanto I, Price P. HIV patients, healthy aging and transplant recipients can reveal the hidden footprints of CMV. Clin Immunol 2017; 187:107-112. [PMID: 29108855 DOI: 10.1016/j.clim.2017.11.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 08/24/2017] [Accepted: 11/01/2017] [Indexed: 12/13/2022]
Abstract
Cytomegalovirus (CMV) is a β-herpesvirus. Latent infections are common in all populations. However age-associated increases in levels of CMV-reactive antibody are testament to repeated reactivations and periods of viral replication. CMV has been associated with several diseases of aging, including vasculopathy and neurocognitive impairment. These conditions occur at a younger age in persons with particularly high burdens of CMV - transplant recipients and people living with HIV. Here we define the "clinical footprints" as immunopathologies triggered by CMV that develop over many years. A high burden of CMV also drives accumulation of multifunctional terminally-differentiated αβ T-cells, a novel population of Vδ2- γδ T-cells, and a population of CD56lo NK cells lacking a key regulatory molecule. An understanding of these "immunological footprints" of CMV may reveal how they collectively promote the "clinical footprints" of the virus. This is explored here in transplant recipients, HIV patients and healthy aging.
Collapse
Affiliation(s)
- Shelley Waters
- School of Biomedical Science, Curtin University, Bentley, Australia
| | - Emily Brook
- School of Biomedical Science, Curtin University, Bentley, Australia
| | - Silvia Lee
- School of Biomedical Science, Curtin University, Bentley, Australia; Department of Microbiology, Pathwest Laboratory Medicine, Fiona Stanley Hospital, Australia
| | - Riwanti Estiasari
- Faculty of Medicine, Universitas Indonesia and Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Ibnu Ariyanto
- Faculty of Medicine, Universitas Indonesia and Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Patricia Price
- School of Biomedical Science, Curtin University, Bentley, Australia; Faculty of Medicine, Universitas Indonesia and Cipto Mangunkusumo Hospital, Jakarta, Indonesia.
| |
Collapse
|
15
|
Sabins NC, Chornoguz O, Leander K, Kaplan F, Carter R, Kinder M, Bachman K, Verona R, Shen S, Bhargava V, Santulli-Marotto S. TIM-3 Engagement Promotes Effector Memory T Cell Differentiation of Human Antigen-Specific CD8 T Cells by Activating mTORC1. THE JOURNAL OF IMMUNOLOGY 2017; 199:4091-4102. [PMID: 29127145 DOI: 10.4049/jimmunol.1701030] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 10/16/2017] [Indexed: 01/19/2023]
Abstract
T cell expression of TIM-3 following Ag encounter has been associated with a continuum of functional states ranging from effector memory T cells to exhaustion. We have designed an in vitro culture system to specifically address the impact of anti-TIM-3/TIM-3 engagement on human Ag-specific CD8 T cells during a normal response to Ag and found that anti-TIM-3 treatment enhances T cell function. In our in vitro T cell culture system, MART1-specific CD8 T cells were expanded from healthy donors using artificial APCs. To ensure that the T cells were the only source of TIM-3, cells were rechallenged with peptide-loaded artificial APCs in the presence of anti-TIM-3 Ab. In these conditions, anti-TIM-3 treatment promotes generation of effector T cells as shown by acquisition of an activated phenotype, increased cytokine production, enhanced proliferation, and a transcription program associated with T cell differentiation. Activation of mTORC1 has been previously demonstrated to enhance CD8 T cell effector function and differentiation. Anti-TIM-3 drives CD8 T cell differentiation through activation of the mTORC1 as evidenced by increased levels of phosphorylated S6 protein and rhebl1 transcript. Altogether these findings suggest that anti-TIM-3, together with Ag, drives differentiation in favor of effector T cells via the activation of mTOR pathway. To our knowledge, this is the first report demonstrating that TIM-3 engagement during Ag stimulation directly influences T cell differentiation through mTORC1.
Collapse
Affiliation(s)
- Nina Chi Sabins
- Janssen Biotherapeutics, Janssen Research and Development, Spring House, PA 19477
| | - Olesya Chornoguz
- Janssen Biotherapeutics, Janssen Research and Development, Spring House, PA 19477
| | - Karen Leander
- Janssen Biotherapeutics, Janssen Research and Development, Spring House, PA 19477
| | - Fred Kaplan
- Oncology, Janssen Research and Development, Spring House, PA 19477
| | - Richard Carter
- Janssen Biotherapeutics, Janssen Research and Development, Spring House, PA 19477
| | - Michelle Kinder
- Oncology, Janssen Research and Development, Spring House, PA 19477
| | - Kurtis Bachman
- Oncology, Janssen Research and Development, Spring House, PA 19477
| | - Raluca Verona
- Oncology, Janssen Research and Development, Spring House, PA 19477
| | - Shixue Shen
- Oncology, Janssen Research and Development, Spring House, PA 19477
| | - Vipul Bhargava
- Computational and Systems Biology, Janssen Research and Development, Spring House, PA 19477; and
| | | |
Collapse
|
16
|
Dekeyser M, Ladrière M, Audonnet S, Frimat L, De Carvalho Bittencourt M. An Early Immediate Early Protein IE-1-Specific T-Cell Polyfunctionality Is Associated With a Better Control of Cytomegalovirus Reactivation in Kidney Transplantation. Kidney Int Rep 2017; 2:486-492. [PMID: 29142976 PMCID: PMC5678683 DOI: 10.1016/j.ekir.2017.02.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 02/21/2017] [Accepted: 02/22/2017] [Indexed: 10/25/2022] Open
Affiliation(s)
- Manon Dekeyser
- Department of Nephrology and Kidney Transplantation, CHRU Nancy, Nancy, France
| | - Marc Ladrière
- Department of Nephrology and Kidney Transplantation, CHRU Nancy, Nancy, France
| | - Sandra Audonnet
- Laboratory of Immunology, Nancytomique, Pôle Laboratoires, CHRU Nancy, Nancy, France
| | - Luc Frimat
- Department of Nephrology and Kidney Transplantation, CHRU Nancy, Nancy, France
| | | |
Collapse
|
17
|
Nada MH, Wang H, Workalemahu G, Tanaka Y, Morita CT. Enhancing adoptive cancer immunotherapy with Vγ2Vδ2 T cells through pulse zoledronate stimulation. J Immunother Cancer 2017; 5:9. [PMID: 28239463 PMCID: PMC5319075 DOI: 10.1186/s40425-017-0209-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 01/06/2017] [Indexed: 01/14/2023] Open
Abstract
Background Human γδ T cells expressing Vγ2Vδ2 T cell receptors monitor foreign- and self-prenyl pyrophosphate metabolites in isoprenoid biosynthesis to mediate immunity to microbes and tumors. Adoptive immunotherapy with Vγ2Vδ2 T cells has been used to treat cancer patients with partial and complete remissions. Most clinical trials and preclinical studies have used continuous zoledronate exposure to expand Vγ2Vδ2 cells where zoledronate is slowly diluted over the course of the culture. Zoledronate inhibits farnesyl diphosphate synthase (FDPS) in monocytes causing isopentenyl pyrophosphate to accumulate that then stimulates Vγ2Vδ2 cells. Because zoledronate inhibition of FDPS is also toxic for T cells, we hypothesized that a short period of exposure would reduce T cell toxicity but still be sufficient for monocytes uptake. Additionally, IL-15 increases the anti-tumor activity of murine αβ T cells in mice but its effect on the in vivo anti-tumor activity of human Vγ2Vδ2 cells has not been assessed. Methods Human Vγ2Vδ2 T cells were expanded by pulse or continuous zoledronate stimulation with IL-2 or IL-15. Expanded Vγ2Vδ2 cells were tested for their expression of effector molecules and killing of tumor cells as well as their in vivo control of human prostate cancer tumors in immunodeficient NSG mice. Results Pulse zoledronate stimulation with either IL-2 or IL-15 resulted in more uniform expansion of Vγ2Vδ2 cells with higher purity and cell numbers as compared with continuous exposure. The Vγ2Vδ2 cells had higher levels of CD107a and perforin and increased tumor cytotoxicity. Adoptive immunotherapy with Vγ2Vδ2 cells derived by pulse stimulation controlled human PC-3 prostate cancer tumors in NSG mice significantly better than those derived by continuous stimulation, halting tumor growth. Although pulse zoledronate stimulation with IL-15 preserved early memory subsets, adoptive immunotherapy with IL-15-derived Vγ2Vδ2 cells equally inhibited PC-3 tumor growth as those derived with IL-2. Conclusions Pulse zoledronate stimulation maximizes the purity, quantity, and quality of expanded Vγ2Vδ2 cells for adoptive immunotherapy but there is no advantage to using IL-15 over IL-2 in our humanized mouse model. Pulse zoledronate stimulation is a simple modification to existing protocols that will enhance the effectiveness of adoptively transferred Vγ2Vδ2 cells by increasing their numbers and anti-tumor activity. Electronic supplementary material The online version of this article (doi:10.1186/s40425-017-0209-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mohanad H Nada
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242 USA.,Department of Veterans Affairs, Iowa City Health Care System, Iowa City, IA 52246 USA.,Interdisciplinary Graduate Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242 USA.,Department of Pathology, College of Medicine, Tikrit University, Tikrit, Iraq
| | - Hong Wang
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242 USA.,Department of Veterans Affairs, Iowa City Health Care System, Iowa City, IA 52246 USA
| | - Grefachew Workalemahu
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242 USA.,Department of Veterans Affairs, Iowa City Health Care System, Iowa City, IA 52246 USA
| | - Yoshimasa Tanaka
- Center for Bioinformatics and Molecular Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan
| | - Craig T Morita
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242 USA.,Department of Veterans Affairs, Iowa City Health Care System, Iowa City, IA 52246 USA.,Interdisciplinary Graduate Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242 USA
| |
Collapse
|
18
|
Khairallah C, Déchanet-Merville J, Capone M. γδ T Cell-Mediated Immunity to Cytomegalovirus Infection. Front Immunol 2017; 8:105. [PMID: 28232834 PMCID: PMC5298998 DOI: 10.3389/fimmu.2017.00105] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 01/20/2017] [Indexed: 12/28/2022] Open
Abstract
γδ T lymphocytes are unconventional immune cells, which have both innate- and adaptive-like features allowing them to respond to a wide spectrum of pathogens. For many years, we and others have reported on the role of these cells in the immune response to human cytomegalovirus in transplant patients, pregnant women, neonates, immunodeficient children, and healthy people. Indeed, and as described for CD8+ T cells, CMV infection leaves a specific imprint on the γδ T cell compartment: (i) driving a long-lasting expansion of oligoclonal γδ T cells in the blood of seropositive individuals, (ii) inducing their differentiation into effector/memory cells expressing a TEMRA phenotype, and (iii) enhancing their antiviral effector functions (i.e., cytotoxicity and IFNγ production). Recently, two studies using murine CMV (MCMV) have corroborated and extended these observations. In particular, they have illustrated the ability of adoptively transferred MCMV-induced γδ T cells to protect immune-deficient mice against virus-induced death. In vivo, expansion of γδ T cells is associated with the clearance of CMV infection as well as with reduced cancer occurrence or leukemia relapse risk in kidney transplant patients and allogeneic stem cell recipients, respectively. Taken together, all these studies show that γδ T cells are important immune effectors against CMV and cancer, which are life-threatening diseases affecting transplant recipients. The ability of CMV-induced γδ T cells to act independently of other immune cells opens the door to the development of novel cellular immunotherapies that could be particularly beneficial for immunocompromised transplant recipients.
Collapse
Affiliation(s)
| | | | - Myriam Capone
- Immunoconcept, CNRS UMR 5164, Bordeaux University, Bordeaux, France
| |
Collapse
|
19
|
van Aalderen MC, Remmerswaal EBM, Heutinck KM, ten Brinke A, Feltkamp MCW, van der Weerd NC, van der Pant KAMI, Bemelman FJ, van Lier RAW, ten Berge IJM. Clinically Relevant Reactivation of Polyomavirus BK (BKPyV) in HLA-A02-Positive Renal Transplant Recipients Is Associated with Impaired Effector-Memory Differentiation of BKPyV-Specific CD8+ T Cells. PLoS Pathog 2016; 12:e1005903. [PMID: 27723787 PMCID: PMC5056763 DOI: 10.1371/journal.ppat.1005903] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/29/2016] [Indexed: 12/24/2022] Open
Abstract
Polyomavirus BK (BKPyV) frequently reactivates in immunosuppressed renal transplant recipients (RTRs) and may lead to graft loss due to BKPyV-induced interstitial nephritis (BKVN). Little is known on the differentiation of CD8+ T cells targeting BKPyV in RTRs. Here we investigated whether BKPyV-specific CD8+ T cell differentiation differs in RTRs with varying degrees of BKPyV reactivation and/or BKVN. Using combinatorial encoding with tetramers carrying BKPyV major capsid protein (VP1) and large T antigen protein (LTAG) epitopes, we investigated CD8+ T cell responses to BKPyV in longitudinally obtained PBMC samples from 46 HLA-A02-positive RTRs and 20 healthy adults. We were also able to isolate BKPyV-specific CD8+ T cells from five renal allografts, two of which were affected by BKVN. Before transplantation, BKPyV-specific CD8+ T cells targeting VP1 and LTAG epitopes appeared predominantly as central-memory and CD27+/CD28+ effector-memory (TEM), and naïve-like PD-1-expressing cells, respectively. After viral reactivation, BKPyV-specific CD8+ T cells assumed CD28− TEM and TEMRA states in patients who were able to control BKPyV, whereas differentiation lagged behind in patients with severe viral reactivation or BKVN. Furthermore, VP1-specific CD69+/CD103+ tissue-resident memory (TRM) cells accumulated in BKVN-affected allografts but lacked signs of effector differentiation. In contrast, granzyme B-expressing effector cells were detected in allografts not affected by BKVN. In conclusion, effector-memory differentiation of BKPyV-specific CD8+ T cells in patients with high viral load or BKVN is impaired. Further characterization of the specific mechanisms behind this altered cellular differentiation is necessary to develop therapies that can prevent the emergence of BKVN. In immunosuppressed renal transplant recipients (RTRs), BKPyV frequently reactivates from latency and may cause severe interstitial nephritis in the allograft (BKVN). Not only is there no effective treatment, it also not understood why BKVN arises in some RTRs but not in all. In the current study we investigated populations of CD8+ T cells targeting epitopes from structural and non-structural BKPyV proteins in RTRs over the course of transplantation. In contrast to RTRs who suffered from self-limiting reactivation of BKPyV, patients who developed severe viral reactivation and BKVN were found to have BKPyV-specific CD8+ T cells which did not, or less often differentiate into CD28− effector-memory cells during viral reactivation. Moreover, virus-specific CD8+ T cell activation and differentiation was not only impaired in the circulation, but possibly also in BKVN-affected renal allografts. In contrast to the CD8+ T cells in kidneys from three patients who did not develop BKVN, T cells in two BKVN-affected kidneys did not display typical cytotoxic effector traits. These findings suggest that impaired BKPyV-specific CD8+ T cell maturation in response to viral reactivation, possibly owing to inter-individual differences in sensitivity to immunosuppressive medication or to certain viral quasispecies, underlies the emergence of severe viral reactivation and BKVN.
Collapse
Affiliation(s)
- Michiel C. van Aalderen
- Department of Experimental Immunology, Amsterdam, the Netherlands
- Renal Transplant Unit, Division of Internal Medicine, Academic Medical Center, Amsterdam, the Netherlands
- * E-mail:
| | - Ester B. M. Remmerswaal
- Department of Experimental Immunology, Amsterdam, the Netherlands
- Renal Transplant Unit, Division of Internal Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | - Kirstin M. Heutinck
- Department of Experimental Immunology, Amsterdam, the Netherlands
- Renal Transplant Unit, Division of Internal Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | - Anja ten Brinke
- Sanquin Blood Supply Foundation and Landsteiner laboratory, Amsterdam, the Netherlands
| | - Mariet C. W. Feltkamp
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Neelke C. van der Weerd
- Renal Transplant Unit, Division of Internal Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | | | - Frederike J. Bemelman
- Renal Transplant Unit, Division of Internal Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | - René A. W. van Lier
- Sanquin Blood Supply Foundation and Landsteiner laboratory, Amsterdam, the Netherlands
| | - Ineke J. M. ten Berge
- Department of Experimental Immunology, Amsterdam, the Netherlands
- Renal Transplant Unit, Division of Internal Medicine, Academic Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
20
|
Bruno L, Cortese M, Monda G, Gentile M, Calò S, Schiavetti F, Zedda L, Cattaneo E, Piccioli D, Schaefer M, Notomista E, Maione D, Carfì A, Merola M, Uematsu Y. Human cytomegalovirus pUL10 interacts with leukocytes and impairs TCR-mediated T-cell activation. Immunol Cell Biol 2016; 94:849-860. [PMID: 27192938 DOI: 10.1038/icb.2016.49] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 05/11/2016] [Accepted: 05/11/2016] [Indexed: 01/27/2023]
Abstract
Human cytomegalovirus (HCMV) is known to exert suppressive effects on the host immune system through expression of various viral genes, thus directly and indirectly affecting antiviral immunity of the infected individuals. We report here that HCMV UL10 encodes a protein (pUL10) with immunosuppressive properties. UL10 has been classified as a member of the HCMV RL11 gene family. Although pUL10 is known to be dispensable for viral replication in cultured cells, its amino-acid sequence is well conserved among different HCMV isolates, suggesting that the protein has a crucial role in viral survival in the host environment. We show that pUL10 is cleaved from the cell surface of fibroblasts as well as epithelial cells and interacts with a cellular receptor ubiquitously expressed on the surface of human leukocytes, demonstrated by ex vivo cell-based assays and flow cytometric analyses on both lymphoid cell lines and primary blood cells. Furthermore, preincubation of peripheral blood mononuclear cells with purified pUL10 ectodomain results in significantly impaired proliferation and substantially reduced pro-inflammatory cytokine production, in particular in CD4+ T cells upon in vitro T-cell stimulation. The inhibitory effect of pUL10 is also observed on antigen receptor-mediated intracellular tyrosine phosphorylation in a T-cell line. Based on these observations, we suggest that pUL10 is a newly identified immunomodulatory protein encoded by HCMV. Further elucidation of interactions between pUL10 and the host immune system during HCMV may contribute to finding ways towards new therapies for HCMV infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Eugenio Notomista
- Department of Biology, University of Naples 'Federico II', Naples, Italy
| | | | | | - Marcello Merola
- Research, GSK Vaccines, Siena, Italy.,Department of Biology, University of Naples 'Federico II', Naples, Italy
| | | |
Collapse
|
21
|
Heutinck KM, Yong SL, Tonneijck L, van den Heuvel H, van der Weerd NC, van der Pant KAMI, Bemelman FJ, Claas FHJ, Ten Berge IJM. Virus-Specific CD8(+) T Cells Cross-Reactive to Donor-Alloantigen Are Transiently Present in the Circulation of Kidney Transplant Recipients Infected With CMV and/or EBV. Am J Transplant 2016; 16:1480-91. [PMID: 26603974 DOI: 10.1111/ajt.13618] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/05/2015] [Accepted: 10/24/2015] [Indexed: 01/25/2023]
Abstract
T cells play a dual role in transplantation: They mediate transplant rejection and are crucial for virus control. Memory T cells generated in response to pathogens can cross-react to alloantigen, a phenomenon called heterologous immunity. Virus-specific CD8(+) T cells cross-reacting to donor-alloantigen might affect alloimmune responses and hamper tolerance induction following transplantation. Here, we longitudinally studied these cross-reactive cells in peripheral blood of 25 kidney transplant recipients with a cytomegalovirus and/or Epstein-Barr virus infection. Cross-reactive T cells were identified by flow cytometry as virus-specific T cells that proliferate in response to donor cells in a mixed-lymphocyte reaction. In 13 of 25 patients, we found cross-reactivity to donor cells for at least 1 viral epitope before (n = 7) and/or after transplantation (n = 8). Cross-reactive T cells were transiently present in the circulation, and their precursor frequency did not increase following transplantation or viral infection. Cross-reactive T cells expressed interferon-γ and CD107a in response to both alloantigen and viral peptide and resembled virus-specific T cells in phenotype and function. Their presence was not associated with impaired renal function, proteinuria, or rejection. In conclusion, virus-specific T cells that cross-react to donor-alloantigen are transiently detectable in the circulation of kidney transplant recipients.
Collapse
Affiliation(s)
- K M Heutinck
- Department of Experimental Immunology, Academic Medical Center, Amsterdam, The Netherlands.,Renal Transplant Unit, Department of Nephrology, Division of Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - S L Yong
- Department of Experimental Immunology, Academic Medical Center, Amsterdam, The Netherlands.,Renal Transplant Unit, Department of Nephrology, Division of Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - L Tonneijck
- Department of Experimental Immunology, Academic Medical Center, Amsterdam, The Netherlands
| | - H van den Heuvel
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - N C van der Weerd
- Renal Transplant Unit, Department of Nephrology, Division of Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - K A M I van der Pant
- Renal Transplant Unit, Department of Nephrology, Division of Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - F J Bemelman
- Renal Transplant Unit, Department of Nephrology, Division of Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - F H J Claas
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - I J M Ten Berge
- Renal Transplant Unit, Department of Nephrology, Division of Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Setoguchi R. IL-15 boosts the function and migration of human terminally differentiated CD8+T cells by inducing a unique gene signature. Int Immunol 2016; 28:293-305. [DOI: 10.1093/intimm/dxw004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/03/2016] [Indexed: 12/13/2022] Open
|
23
|
Braun J, Frentsch M, Thiel A. Hobit and human effector T-cell differentiation: The beginning of a long journey. Eur J Immunol 2016; 45:2762-5. [PMID: 26440905 DOI: 10.1002/eji.201545959] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 09/04/2015] [Accepted: 09/07/2015] [Indexed: 01/06/2023]
Abstract
Besides growing plants, eating a lot, and drinking beer, Tolkien's Hobbits enjoy maintaining a quiet state. Regarding the latter, the name chosen for a recently discovered transcription factor seems to be unintentionally appropriate. The zinc finger protein ZNF683 was originally named "Hobit" for Homolog of Blimp-1 in T cells. In this issue of the European Journal of Immunology, Braga et al. [Eur. J. Immunol. 2015. 45: 2945-2958] demonstrate that in humans, Hobit is almost exclusively expressed in effector T cells, in particular in quiescent and long-lived effector-type CD8(+) T cells. Hobit may initially appear as another "player" in the quest for transcription factors guiding T-cell differentiation; the discoveries of T-bet, Eomes, Blimp-1, and others have significantly contributed to our understanding of how this process is tightly regulated. However, Hobit may be special--the currently available results suggest substantial differences in Hobit's regulatory functions between mice and humans, such as expression patterns and IFN-γ regulation. And it may turn out that Hobit's function in human T cells is highly adapted to lifelong, periodic challenges with varying, physiological doses of pathogens. Thus, the new study about Hobit in human T cells may be the beginning of a long journey.
Collapse
Affiliation(s)
- Julian Braun
- Regenerative Immunology and Aging, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marco Frentsch
- Regenerative Immunology and Aging, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Thiel
- Regenerative Immunology and Aging, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
24
|
Vieira Braga FA, Hertoghs KML, van Lier RAW, van Gisbergen KPJM. Molecular characterization of HCMV-specific immune responses: Parallels between CD8(+) T cells, CD4(+) T cells, and NK cells. Eur J Immunol 2015; 45:2433-45. [PMID: 26228786 DOI: 10.1002/eji.201545495] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 07/15/2015] [Accepted: 07/28/2015] [Indexed: 11/07/2022]
Abstract
CD8(+) T cells are important for immunity against human cytomegalovirus (HCMV). The HCMV-specific CD8(+) T-cell response is characterized by the accumulation of terminally differentiated effector cells that have downregulated the costimulatory molecules CD27 and CD28. These HCMV-specific CD8(+) T cells maintain high levels of cytotoxic molecules such as granzyme B and rapidly produce the inflammatory cytokine IFN-γ upon activation. Remarkably, HCMV-specific CD8(+) T cells are able to persist long term as fully functional effector cells, suggesting a unique differentiation pathway that is distinct from the formation of memory CD8(+) T cells after infection with acute viruses. In this review, we aim to highlight the most recent developments in HCMV-specific CD8(+) T-cell differentiation, maintenance, tissue distribution, metabolism and function. HCMV also induces the differentiation of effector CD4(+) T cells and NK cells, which share characteristics with HCMV-specific CD8(+) T cells. We propose that the overlap in differentiation of NK cells, CD4(+) and CD8(+) T cells after HCMV infection may be regulated by a shared transcriptional machinery. A better understanding of the molecular framework of HCMV-specific CD8(+) T-cell responses may benefit vaccine design, as these cells uniquely combine the capacity to rapidly respond to infection with long-term survival.
Collapse
Affiliation(s)
- Felipe A Vieira Braga
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory AMC/UvA, Amsterdam, The Netherlands
| | - Kirsten M L Hertoghs
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory AMC/UvA, Amsterdam, The Netherlands
| | - René A W van Lier
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory AMC/UvA, Amsterdam, The Netherlands
| | - Klaas P J M van Gisbergen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory AMC/UvA, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Infection history determines the differentiation state of human CD8+ T cells. J Virol 2015; 89:5110-23. [PMID: 25717102 DOI: 10.1128/jvi.03478-14] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 02/17/2015] [Indexed: 01/17/2023] Open
Abstract
UNLABELLED After the resolution of the acute phase of infection, otherwise quiescent antigen-experienced CD8(+) T cells confer rapid protection upon reinfection with viral pathogens or, in the case of persistent viruses, help to maintain control of the infection. Depending on the type of virus, antigen-specific CD8(+) T cells have distinct traits, ranging from typical memory cell properties in the case of rapidly cleared viruses to immediate effector functions for persistent viruses. We here show that both the differentiation stage, defined by the expression of cell surface markers, such as CD45RA, CCR7, CD28, and CD27, and distinct expression levels of T-bet and eomesodermin (Eomes) predict the functional profile of antigen-experienced CD8(+) T cells. Furthermore, virus-specific CD8(+) T cells targeting different respiratory syncytial virus-, influenza A virus-, Epstein-Barr virus (EBV)-, human cytomegalovirus (hCMV)-, and HIV-1-specific epitopes adopt distinct T-bet and Eomes expression patterns that appear to be installed early during the primary response. Importantly, the associations between surface phenotype, T-bet/Eomes expression levels, and the expression of markers that predict CD8(+) T-cell function change according to viral infection history, particularly against the background of HIV-1 and, to lesser extent, of human cytomegalovirus and/or Epstein-Barr virus infection. Thus, the functionality of human antigen-experienced CD8(+) T cells follows at least two dimensions, one outlined by the surface phenotype and another by the T-bet/Eomes expression levels, which are determined by previous or persistent viral challenges. IMPORTANCE Functional human CD8(+) T-cell subsets have been defined using surface markers like CD45RA, CCR7, CD28, and CD27. However, the induction of function-defining traits, like granzyme B expression, is controlled by transcription factors like T-bet and Eomes. Here, we describe how T-bet and Eomes levels distinctly relate to the expression of molecules predictive for CD8(+) T-cell function in a surface phenotype-independent manner. Importantly, we found that central memory and effector memory CD8(+) T-cell subsets differentially express T-bet, Eomes, and molecules predictive for function according to viral infection history, particularly so in the context of HIV-1 infection and, to lesser extent, of latent EBV- and/or hCMV-infected, otherwise healthy adults. Finally, we show that the distinct phenotypes and T-bet/Eomes levels of different virus-specific CD8(+) T-cell populations are imprinted early during the acute phase of primary infection in vivo. These findings broaden our understanding of CD8(+) T-cell differentiation.
Collapse
|
26
|
Quinn M, Turula H, Tandon M, Deslouches B, Moghbeli T, Snyder CM. Memory T cells specific for murine cytomegalovirus re-emerge after multiple challenges and recapitulate immunity in various adoptive transfer scenarios. THE JOURNAL OF IMMUNOLOGY 2015; 194:1726-1736. [PMID: 25595792 DOI: 10.4049/jimmunol.1402757] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Reconstitution of CMV-specific immunity after transplant remains a primary clinical objective to prevent CMV disease, and adoptive immunotherapy of CMV-specific T cells can be an effective therapeutic approach. Because of viral persistence, most CMV-specific CD8(+) T cells become terminally differentiated effector phenotype CD8(+) T cells (TEFF). A minor subset retains a memory-like phenotype (memory phenotype CD8(+) T cells [TM]), but it is unknown whether these cells retain memory function or persist over time. Interestingly, recent studies suggest that CMV-specific CD8(+) T cells with different phenotypes have different abilities to reconstitute sustained immunity after transfer. The immunology of human CMV infections is reflected in the murine CMV (MCMV) model. We found that human CMV- and MCMV-specific T cells displayed shared genetic programs, validating the MCMV model for studies of CMV-specific T cells in vivo. The MCMV-specific TM population was stable over time and retained a proliferative capacity that was vastly superior to TEFF. Strikingly, after transfer, TM established sustained and diverse T cell populations even after multiple challenges. Although both TEFF and TM could protect Rag(-/-) mice, only TM persisted after transfer into immune replete, latently infected recipients and responded if recipient immunity was lost. Interestingly, transferred TM did not expand until recipient immunity was lost, supporting that competition limits the Ag stimulation of TM. Ultimately, these data show that CMV-specific TM retain memory function during MCMV infection and can re-establish CMV immunity when necessary. Thus, TM may be a critical component for consistent, long-term adoptive immunotherapy success.
Collapse
Affiliation(s)
- Michael Quinn
- Department of Immunology and Microbial Pathogenesis, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Holly Turula
- Department of Immunology and Microbial Pathogenesis, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Mayank Tandon
- Department of Immunology and Microbial Pathogenesis, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Berthony Deslouches
- Department of Immunology and Microbial Pathogenesis, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Toktam Moghbeli
- Department of Immunology and Microbial Pathogenesis, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Christopher M Snyder
- Department of Immunology and Microbial Pathogenesis, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
27
|
ten Berge IJ, van Lier RA. The interaction between cytomegalovirus and the human immune system. Immunol Lett 2014; 162:141-4. [DOI: 10.1016/j.imlet.2014.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
28
|
ten Berge IJM, Bemelman FJ. Basic and clinical research in the AMC Renal Transplant Unit. Transpl Immunol 2014; 31:210-2. [PMID: 25240734 DOI: 10.1016/j.trim.2014.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 09/08/2014] [Indexed: 10/24/2022]
Abstract
Our research is aimed at characterization of the antiviral and allo-immune responses in kidney transplant recipients. In particular, we are interested in the differentiation, effector function and memory formation of T cells specific for the latent viruses cytomegalovirus (CMV) and BK virus (BKV) and in the impact of virus-specific immune responses on alloimmunity. Furthermore, we perform studies towards the most optimal, tailor-made immunosuppressive drug regimen and towards the impact of immunosuppression on infectious diseases; cognitive function and cardiovascular status of kidney transplant recipients.
Collapse
Affiliation(s)
- Ineke J M ten Berge
- Renal Transplant Unit, Department of Internal Medicine, Academic Medical Centre, Amsterdam, The Netherlands.
| | - Fréderike J Bemelman
- Renal Transplant Unit, Department of Internal Medicine, Academic Medical Centre, Amsterdam, The Netherlands
| |
Collapse
|
29
|
Rodrigues L, Bonorino C. Role of IL-15 and IL-21 in viral immunity: applications for vaccines and therapies. Expert Rev Vaccines 2014; 8:167-77. [DOI: 10.1586/14760584.8.2.167] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
30
|
CMV-specific CD8+ T-cell function is not impaired in chronic lymphocytic leukemia. Blood 2013; 123:717-24. [PMID: 24246502 DOI: 10.1182/blood-2013-08-518183] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
In chronic lymphocytic leukemia (CLL), CD8(+) T cells exhibit features of exhaustion and impaired functionality. Yet, reactivations of latent viruses such as cytomegalovirus (CMV) are uncommon in untreated CLL, suggesting that antiviral responses are uncompromised. We analyzed phenotypical and functional characteristics of CMV-specific CD8(+) T cells in CLL patients in comparison with age-matched healthy controls (HCs). Despite increased expression of the inhibitory receptors PD1, CD160, and CD244 on total CD8(+) T cells in CLL, expression levels of these markers were decreased on CMV-tetramer(+)CD8(+) T cells. Second, cytokine production upon stimulation with both phorbol 12-myristate 13-acetate/ionomycin and CMV-peptide-loaded antigen-presenting cells was intact in CMV-tetramer(+)CD8(+) T cells. Third, CMV-tetramer(+)CD8(+) T cells of CLL patients and HCs were equally effective in killing CMV-peptide-loaded target cells. Finally, quantitative imaging flow cytometry revealed that the proportion of CD8(+) T cells forming immunologic synapses with CMV-peptide-loaded B cells was intact. In conclusion, despite evidence for global T-cell dysfunction in CLL, we show here that CLL-derived CMV-specific CD8(+) T cells display lower expression of exhaustion markers and are functionally intact. These data indicate that the changes in the T-cell compartment in CLL may be more heterogeneous than presently assumed.
Collapse
|
31
|
Pulendran B, Oh JZ, Nakaya HI, Ravindran R, Kazmin DA. Immunity to viruses: learning from successful human vaccines. Immunol Rev 2013; 255:243-55. [PMID: 23947360 PMCID: PMC3748616 DOI: 10.1111/imr.12099] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
For more than a century, immunologists and vaccinologists have existed in parallel universes. Immunologists have for long reveled in using 'model antigens', such as chicken egg ovalbumin or nitrophenyl haptens, to study immune responses in model organisms such as mice. Such studies have yielded many seminal insights about the mechanisms of immune regulation, but their relevance to humans has been questioned. In another universe, vaccinologists have relied on human clinical trials to assess vaccine efficacy, but have done little to take advantage of such trials for studying the nature of immune responses to vaccination. The human model provides a nexus between these two universes, and recent studies have begun to use this model to study the molecular profile of innate and adaptive responses to vaccination. Such 'systems vaccinology' studies are beginning to provide mechanistic insights about innate and adaptive immunity in humans. Here, we present an overview of such studies, with particular examples from studies with the yellow fever and the seasonal influenza vaccines. Vaccination with the yellow fever vaccine causes a systemic acute viral infection and thus provides an attractive model to study innate and adaptive responses to a primary viral challenge. Vaccination with the live attenuated influenza vaccine causes a localized acute viral infection in mucosal tissues and induces a recall response, since most vaccinees have had prior exposure to influenza, and thus provides a unique opportunity to study innate and antigen-specific memory responses in mucosal tissues and in the blood. Vaccination with the inactivated influenza vaccine offers a model to study immune responses to an inactivated immunogen. Studies with these and other vaccines are beginning to reunite the estranged fields of immunology and vaccinology, yielding unexpected insights about mechanisms of viral immunity. Vaccines that have been proven to be of immense benefit in saving lives offer us a new fringe benefit: lessons in viral immunology.
Collapse
|
32
|
Smith C, Khanna R. Immune regulation of human herpesviruses and its implications for human transplantation. Am J Transplant 2013; 13 Suppl 3:9-23; quiz 23. [PMID: 23347211 DOI: 10.1111/ajt.12005] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 07/03/2012] [Accepted: 07/16/2012] [Indexed: 01/25/2023]
Abstract
Human herpesviruses including cytomegalovirus, Epstein-Barr virus, HHV6, HHV7, HHV8, Herpes simplex virus (HSV)-1 and HSV-2 and varicella zoster virus (VZV) have developed an intricate relationship with the human immune system. This is characterized by the interplay between viral immune evasion mechanisms that promote the establishment of a lifelong persistent infection and the induction of a broad humoral and cellular immune response, which prevents the establishment of viral disease. Understanding the immune parameters that control herpesvirus infection, and the strategies the viruses use to evade immune recognition, has been critical in understanding why immunological dysfunction in transplant patients can lead to disease, and in the development of immunological strategies to prevent and control herpesvirus associated diseases.
Collapse
Affiliation(s)
- C Smith
- Australian Centre for Vaccine Development, Tumour Immunology Laboratory, Department of Immunology, Queensland Institute of Medical Research, Brisbane, Australia
| | | |
Collapse
|
33
|
Akulian JA, Pipeling MR, John ER, Orens JB, Lechtzin N, McDyer JF. High-quality CMV-specific CD4+ memory is enriched in the lung allograft and is associated with mucosal viral control. Am J Transplant 2013; 13:146-56. [PMID: 23016698 PMCID: PMC3827914 DOI: 10.1111/j.1600-6143.2012.04282.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 08/13/2012] [Accepted: 08/19/2012] [Indexed: 01/25/2023]
Abstract
The maintenance of CMV-specific T cell memory in lung transplant recipients (LTRs) is critical for host defense and allograft durability, particularly in donor(+) /recipient(-) (D(+) R(-) ) individuals who demonstrate increased mortality. We studied CD4(+) and CD8(+) CMV-specific memory responses to phosphoprotein 65 (pp65) in a prospective cohort of 18 D(+) R(-) LTRs, from bronchoalveolar lavage (BAL)-obtained lung mononuclear cells (LMNC) and PBMC. Unexpectedly, pp65-specific CD4(+) and CD8(+) IFN-γ memory responses from LMNC were similar, in contrast to persistent CD8(+) predominance in PBMC. Unlike the pulmonary CD8(+) predominance during acute primary infection, compartmental equalization occurred in the CMV-specific CD8(+) memory pool during chronic infection, whereas CMV-specific CD4(+) memory was enriched in the bronchoalveolar space. Moreover, CMV-specific CD4(+) memory T cells with multifunctional production of IFN-γ, TNF-α, IL-2 and MIP-1β were significantly increased in LMNCs, in contrast to similar intercompartmental CD8(+) memory function. Moreover, the absolute number of CMV-specific CD4(+) IFN-γ(+) memory cells in BAL was significantly increased in LTRs exhibiting viral control compared to those with CMV early antigen positivity. Collectively, these data demonstrate both preferential distribution and functional quality of CMV-specific CD4(+) memory in the lung allograft during chronic infection, and show an important association with CMV mucosal immunity and viral control.
Collapse
Affiliation(s)
- J. A. Akulian
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - M. R. Pipeling
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - E. R. John
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - J. B. Orens
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - N. Lechtzin
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - J. F. McDyer
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD,Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA,Corresponding author: John F. McDyer,
| |
Collapse
|
34
|
Iannello A, Allam O, Samarani S, Ahmad A. [Interleukin-21: a key cytokine for controlling HIV and other chronic viral infections]. Med Sci (Paris) 2012; 28:605-11. [PMID: 22805136 DOI: 10.1051/medsci/2012286013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The differentiation, homeostatic proliferation and effector functions of different immune cells are controlled, to a large extent, by cytokines. Viruses often cause immune response dysfunctions by causing defects in the cytokine networks. The defects are often manifested by altered cytokine secretion and/or responsiveness to the cytokine. Among these cytokines, Interleukin-21 (IL-21) is a relatively recently discovered cytokine, which is mainly produced by CD4(+) T cells in the body, and exerts multiple and pleiotropic effects on various immune cells. Recent studies have shown that the cytokine is indispensable for controlling chronic viral infections. This review summarizes current knowledges concerning the biological effects of this cytokine on different components of the immune system. We also discuss how it contributes toward mounting efficient antiviral immunity and controlling chronic viral infections, especially HIV-1. The IL-1 cytokine represents a novel therapeutic agent for virus-infected patients as well as an adjuvant in antiviral vaccination strategies.
Collapse
Affiliation(s)
- Alexandre Iannello
- Département de microbiologie et d'immunologie, Université de Montréal, Montréal, Québec H3T 1C5, Canada.
| | | | | | | |
Collapse
|
35
|
Cytomegalovirus-induced effector T cells cause endothelial cell damage. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 19:772-9. [PMID: 22398244 DOI: 10.1128/cvi.00011-12] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Human cytomegalovirus (CMV) infection has been linked to inflammatory diseases that involve vascular endothelial cell damage, but definitive proof for a direct cytopathic effect of CMV in these diseases is lacking. CMV infection is associated with a strong increase in both CD4(+) and CD8(+) T cells with constitutive effector functions that can perpetuate systemic inflammation. We investigated whether CMV-induced immune responses could lead to endothelial damage in humans. We found that terminally differentiated effector CD4(+) and CD8(+) T cells, formed during primary CMV infection and maintained throughout latency, express high levels of CX3CR1 and CXCR3. The ligands of these receptors, fractalkine and IP-10, respectively, are expressed by activated endothelial cells. Peripheral blood mononuclear cells (PBMC) stimulated with CMV antigen produced soluble factors that stimulated endothelial cells to produce both chemokines. Finally, effector cells migrated in a fractalkine- and IP-10-dependent fashion to activated endothelial cells and induced apoptosis in endothelial cells that were stimulated by supernatant from CMV-activated PBMC. Our findings offer an explanation for the accumulation of highly differentiated T cells near to the endothelium in CMV-infected individuals that may result in endothelial damage.
Collapse
|
36
|
Smith C, Miles JJ, Khanna R. Advances in direct T-cell alloreactivity: function, avidity, biophysics and structure. Am J Transplant 2012; 12:15-26. [PMID: 22152064 DOI: 10.1111/j.1600-6143.2011.03863.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Although T-cell-based adaptive immunity plays a crucial role in protection against infectious pathogens and uncontrolled outgrowth of malignant cells, a large portion of these T cells are also capable of responding to allogeneic HLA molecules, violating the paradigm of self-major histocompatibility complex (MHC) restriction. Recent studies have provided insights into the mechanisms by which these T cells recognize allogeneic targets. The role of antiviral T cells in direct alloreactivity through peptide-dependent molecular mimicry and alternate peptide-MHC docking modes has emerged as major models for the human alloresponse. Here, we review in depth recent advances in this field and discuss how molecular interactions between T cells and HLA molecules drive the activation of these effector cells and its potential implications for alloreactivity in human transplantation.
Collapse
Affiliation(s)
- C Smith
- Australian Centre for Vaccine Development, Tumour Immunology Laboratory, Queensland Institute of Medical Research, Herston, Brisbane, Australia
| | | | | |
Collapse
|
37
|
Buffered memory: a hypothesis for the maintenance of functional, virus-specific CD8+ T cells during cytomegalovirus infection. Immunol Res 2011; 51:195-204. [DOI: 10.1007/s12026-011-8251-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
38
|
Hertoghs KML, Moerland PD, van Stijn A, Remmerswaal EBM, Yong SL, van de Berg PJEJ, van Ham SM, Baas F, ten Berge IJM, van Lier RAW. Molecular profiling of cytomegalovirus-induced human CD8+ T cell differentiation. J Clin Invest 2010; 120:4077-90. [PMID: 20921622 DOI: 10.1172/jci42758] [Citation(s) in RCA: 161] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Accepted: 08/18/2010] [Indexed: 12/31/2022] Open
Abstract
CD8+ T cells play a critical role in the immune response to viral pathogens. Persistent human cytomegalovirus (HCMV) infection results in a strong increase in the number of virus-specific, quiescent effector-type CD8+ T cells with constitutive cytolytic activity, but the molecular pathways involved in the induction and maintenance of these cells are unknown. We show here that HCMV infection induced acute and lasting changes in the transcriptomes of virus-reactive T cells collected from HCMV-seropositive patients at distinct stages of infection. Enhanced cell cycle and metabolic activity was restricted to the acute phase of the response, but at all stages, HCMV-specific CD8+ T cells expressed the Th1-associated transcription factors T-bet (TBX21) and eomesodermin (EOMES), in parallel with continuous expression of IFNG mRNA and IFN-γ-regulated genes. The cytolytic proteins granzyme B and perforin as well as the fractalkine-binding chemokine receptor CX3CR1 were found in virus-reactive cells throughout the response. During HCMV latency, virus-specific CD8+ T cells lacked the typical features of exhausted cells found in other chronic infections. Persistent effector cell traits together with the permanent changes in chemokine receptor usage of virus-specific, nonexhausted, long-lived CD8+ T cells may be crucial to maintain lifelong protection from HCMV reactivation.
Collapse
Affiliation(s)
- Kirsten M L Hertoghs
- Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Humphreys IR, Lee SW, Jones M, Loewendorf A, Gostick E, Price DA, Benedict CA, Ware CF, Croft M. Biphasic role of 4-1BB in the regulation of mouse cytomegalovirus-specific CD8(+) T cells. Eur J Immunol 2010; 40:2762-8. [PMID: 20722077 PMCID: PMC2967573 DOI: 10.1002/eji.200940256] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Revised: 06/10/2010] [Accepted: 07/19/2010] [Indexed: 12/14/2022]
Abstract
The initial requirement for the emergence of CMV-specific CD8(+) T cells is poorly understood. Mice deficient in the cosignaling TNF superfamily member, 4-1BB, surprisingly developed exaggerated early CD8(+) T-cell responses to mouse CMV (MCMV). CD8(+) T cells directed against acute MCMV epitopes were enhanced, demonstrating that 4-1BB naturally antagonizes these primary populations. Paradoxically, 4-1BB-deficient mice displayed reduced accumulation of memory CD8(+) T cells that expand during chronic/latent infection. Importantly, the canonical TNF-related ligand, 4-1BBL, promoted the accumulation of these memory CD8(+) T cells, whereas suppression of acute CD8(+) T cells was independent of 4-1BBL. These data highlight the dual nature of the 4-1BB/4-1BBL system in mediating both stimulatory and inhibitory cosignaling activities during the generation of anti-MCMV immunity.
Collapse
Affiliation(s)
- Ian R Humphreys
- Division of Molecular Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Jones M, Ladell K, Wynn KK, Stacey MA, Quigley MF, Gostick E, Price DA, Humphreys IR. IL-10 restricts memory T cell inflation during cytomegalovirus infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 185:3583-92. [PMID: 20713884 PMCID: PMC3655265 DOI: 10.4049/jimmunol.1001535] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The beta-herpesvirus CMV induces a substantial and progressive expansion of virus-specific memory CD8 T cells, which protect the host against viral reactivation from latency. In this paper, we report that this expansion, or "inflation," of memory T cells is amplified dramatically during mouse CMV infection of IL-10 knockout (IL-10(-/-)) mice. T cells from IL-10(-/-) mice were oligoclonal, exhibited a highly activated phenotype, expressed antiviral cytokines, and degranulated in response to cognate Ag encounter ex vivo. Moreover, latent viral load was reduced in IL-10(-/-) mice. Importantly, these results were recapitulated by IL-10R blockade during chronic/latent infection of wild-type mice. These data demonstrate that regulatory immune mechanisms can influence CMV-specific T cell memory and suggest a possible rationale for the acquisition of functional IL-10 orthologs by herpesviruses.
Collapse
Affiliation(s)
- Morgan Jones
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Heath Park, CF14 4XN, UK
| | - Kristin Ladell
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Heath Park, CF14 4XN, UK
| | - Katherine K. Wynn
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Heath Park, CF14 4XN, UK
| | - Maria A. Stacey
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Heath Park, CF14 4XN, UK
| | - Máire F. Quigley
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Heath Park, CF14 4XN, UK
| | - Emma Gostick
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Heath Park, CF14 4XN, UK
| | - David A. Price
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Heath Park, CF14 4XN, UK
| | - Ian R. Humphreys
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Heath Park, CF14 4XN, UK
| |
Collapse
|
41
|
Sandalova E, Laccabue D, Boni C, Tan AT, Fink K, Ooi EE, Chua R, Shafaeddin Schreve B, Ferrari C, Bertoletti A. Contribution of herpesvirus specific CD8 T cells to anti-viral T cell response in humans. PLoS Pathog 2010; 6:e1001051. [PMID: 20808900 PMCID: PMC2924358 DOI: 10.1371/journal.ppat.1001051] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Accepted: 07/20/2010] [Indexed: 11/18/2022] Open
Abstract
Herpesviruses infect most humans. Their infections can be associated with pathological conditions and significant changes in T cell repertoire but evidences of symbiotic effects of herpesvirus latency have never been demonstrated. We tested the hypothesis that HCMV and EBV-specific CD8 T cells contribute to the heterologous anti-viral immune response. Volume of activated/proliferating virus-specific and total CD8 T cells was evaluated in 50 patients with acute viral infections: 20 with HBV, 12 with Dengue, 12 with Influenza, 3 with Adenovirus infection and 3 with fevers of unknown etiology. Virus-specific (EBV, HCMV, Influenza) pentamer+ and total CD8 T cells were analyzed for activation (CD38/HLA-DR), proliferation (Ki-67/Bcl-2(low)) and cytokine production. We observed that all acute viral infections trigger an expansion of activated/proliferating CD8 T cells, which differs in size depending on the infection but is invariably inflated by CD8 T cells specific for persistent herpesviruses (HCMV/EBV). CD8 T cells specific for other non-related non persistent viral infection (i.e. Influenza) were not activated. IL-15, which is produced during acute viral infections, is the likely contributing mechanism driving the selective activation of herpesvirus specific CD8 T cells. In addition we were able to show that herpesvirus specific CD8 T cells displayed an increased ability to produce the anti-viral cytokine interferon-gamma during the acute phase of heterologous viral infection. Taken together, these data demonstrated that activated herpesvirus specific CD8 T cells inflate the activated/proliferating CD8 T cells population present during acute viral infections in human and can contribute to the heterologous anti-viral T cell response.
Collapse
Affiliation(s)
- Elena Sandalova
- Singapore Institute for Clinical Sciences, A*STAR, Singapore
| | - Diletta Laccabue
- Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Carolina Boni
- Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Anthony T. Tan
- Singapore Institute for Clinical Sciences, A*STAR, Singapore
| | - Katja Fink
- Singapore Immunology Network, A*STAR, Singapore
| | - Eng Eong Ooi
- Emerging Viral Diseases, Duke – NUS Graduate Medical School, Singapore
| | - Robert Chua
- Emerging Viral Diseases, Duke – NUS Graduate Medical School, Singapore
| | - Bahar Shafaeddin Schreve
- Singapore Institute for Clinical Sciences, A*STAR, Singapore
- Emerging Viral Diseases, Duke – NUS Graduate Medical School, Singapore
| | - Carlo Ferrari
- Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Antonio Bertoletti
- Singapore Institute for Clinical Sciences, A*STAR, Singapore
- Emerging Viral Diseases, Duke – NUS Graduate Medical School, Singapore
- * E-mail:
| |
Collapse
|
42
|
Luo XH, Huang XJ, Liu KY, Xu LP, Liu DH. Protective Immunity Transferred by Infusion of Cytomegalovirus-Specific CD8+ T Cells within Donor Grafts: Its Associations with Cytomegalovirus Reactivation Following Unmanipulated Allogeneic Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant 2010; 16:994-1004. [DOI: 10.1016/j.bbmt.2010.02.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Accepted: 02/05/2010] [Indexed: 12/12/2022]
|
43
|
van de Berg PJEJ, Griffiths SJ, Yong SL, Macaulay R, Bemelman FJ, Jackson S, Henson SM, ten Berge IJM, Akbar AN, van Lier RAW. Cytomegalovirus Infection Reduces Telomere Length of the Circulating T Cell Pool. THE JOURNAL OF IMMUNOLOGY 2010; 184:3417-23. [DOI: 10.4049/jimmunol.0903442] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
44
|
Weinberger B, Welzl K, Herndler-Brandstetter D, Parson W, Grubeck-Loebenstein B. CD28(-)CD8(+) T cells do not contain unique clonotypes and are therefore dispensable. Immunol Lett 2009; 127:27-32. [PMID: 19715728 DOI: 10.1016/j.imlet.2009.08.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Revised: 08/10/2009] [Accepted: 08/18/2009] [Indexed: 11/16/2022]
Abstract
Highly differentiated CD28(-) effector T cells which accumulate in a variety of diseases and also with increasing age contribute to inflammatory processes, limit immunological space and diversity, and are associated with immunological dysfunction and reduced responses to vaccination. Elimination of CD28(-) T cells has been suggested as a measure for immunological rejuvenation but may lead to the loss of important T cell specificities. Using T cells specific for the immunodominant CMV-derived epitope NLVPMVATV as a model, we show that the same clonotypes are present in CD8(+)CD28(+) naïve/early memory and CD8(+)CD28(-) effector T cells. Therefore, CD28(-) cells do not seem to contain clones which are not present in the residual population. The elimination of effector T cells would not lead to the loss of important specificities, as relevant clonotypes could be recruited and propagated from naïve or early memory T cell subsets in the case of exposure to pathogen.
Collapse
Affiliation(s)
- Birgit Weinberger
- Institute for Biomedical Aging Research, Austrian Academy of Sciences, Rennweg 10, 6020 Innsbruck, Austria
| | | | | | | | | |
Collapse
|
45
|
Ebert EC. Interleukin 21 up-regulates perforin-mediated cytotoxic activity of human intra-epithelial lymphocytes. Immunology 2009; 127:206-15. [PMID: 19489126 DOI: 10.1111/j.1365-2567.2008.02941.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Human intra-epithelial lymphocytes (IELs) are predominantly T-cell receptor-alphabeta(+) (TCR-alphabeta(+)) CD8(+) CD45RO(+) memory T cells located between intestinal epithelial cells. They respond to a greater extent to stimulation with interleukin (IL)-15 than to CD3/TCR triggering, suggesting that they react to the cytokine milieu in their local environment rather than to cognate antigen. A newly described member of the gammac cytokine family, IL-21, has potent antitumor effects. As IELs resemble lymphocytes infiltrating neoplastic lesions, their response to IL-21 may be relevant in vivo. Here, IL-21 was shown to increase perforin-mediated cytotoxicity and serine esterase release by IELs. This IL-21-mediated up-regulation occurred without changes in IEL survival or cell division. Interestingly, the effects of IL-21 occurred without increased phosphorylation of signal transducer and activator of transcription (STAT)1, STAT3, STAT4, STAT5, extracellular signal-regulated kinase (ERK), or p38. IL-21 had no effect on Fas ligand (FL)- or tumour necrosis factor-alpha (TNF-alpha)-mediated cytotoxicity, but it down-regulated IL-15-stimulated expression of CD25 and CD94, indicating that it has both positive and negative actions. This functional profile is unique to human IELs, emphasizing that they are a distinct compartment of lymphocytes and that IL-21 may promote their role in tumour immunosurveillance.
Collapse
Affiliation(s)
- Ellen C Ebert
- UMDNJ-Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
| |
Collapse
|
46
|
Brestrich G, Zwinger S, Fischer A, Schmück M, Röhmhild A, Hammer MH, Kurtz A, Uharek L, Knosalla C, Lehmkuhl H, Volk HD, Reinke P. Adoptive T-cell therapy of a lung transplanted patient with severe CMV disease and resistance to antiviral therapy. Am J Transplant 2009; 9:1679-84. [PMID: 19459791 DOI: 10.1111/j.1600-6143.2009.02672.x] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Infections with cytomegalovirus (CMV) can induce severe complications after transplantation, particularly in patients resistant to virostatic therapy. Adoptive transfer of CMV-specific T-cell lines has demonstrated promising results in patients after hematopoietic stem cell transplantation. However, the generation of specific T-cell lines ex vivo and their function in vivo is complicated in solid organ transplant (SOT) recipients. Here, we present the successful adoptive transfer of autologous CMV-specific T cells to a lung transplant recipient with ganciclovir-resistant CMV-pneumonia requiring mechanical ventilation. Infused T cells rapidly expanded in vivo and efficiently inhibited viral replication as confirmed by extensive longitudinal immunological monitoring. After full recovery, the patient was released from the clinic. After 4 weeks, the infection reappeared and persisted at a low level even after a second T-cell infusion. Our experimental data indicate that this could be the consequence of the late differentiated phenotype of the infused T cells and therefore their insufficient longevity in vivo. In summary, our report signifies the high therapeutic potential of adoptive immunotherapy in the treatment of SOT recipients when all other measures show no effect. Further studies have to elucidate the most potent strategies to generate antigen-specific T cells with high functional capacity and robust long-term persistence.
Collapse
Affiliation(s)
- G Brestrich
- Department of Nephrology and Internal Intensive Care, Universitätsmedizin, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
WIESEL MELANIE, WALTON SENTA, RICHTER KIRSTEN, OXENIUS ANNETTE. Virus-specific CD8 T cells: activation, differentiation and memory formation. APMIS 2009; 117:356-81. [DOI: 10.1111/j.1600-0463.2009.02459.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
48
|
Abstract
Neuroblastoma, a solid tumor arising from developing cells of the sympathetic nervous system, is the most common extracranial tumor in children. The prognosis for high-risk neuroblastoma remains poor with conventional treatment, and new approaches are therefore being explored to treat this disease. One such alternative therapy that holds promise is immune therapy. We review here the recent advances in four types of immune therapy-cytokine, vaccine, antibody and cellular therapy-to treat neuroblastoma. We present preclinical research and clinical trials on several promising candidates such as IL-12, dendritic cell vaccines, anti-GD2 antibodies and allogeneic hematopoietic stem cell transplant. An optimal treatment plan for neuroblastoma will most likely involve multimodal approaches and combinations of immune therapies.
Collapse
Affiliation(s)
- Fariba Navid
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105 , USA.
| | | | | |
Collapse
|
49
|
Jagannathan P, Osborne CM, Royce C, Manion MM, Tilton JC, Li L, Fischer S, Hallahan CW, Metcalf JA, McLaughlin M, Pipeling M, McDyer JF, Manley TJ, Meier JL, Altman JD, Hertel L, Davey RT, Connors M, Migueles SA. Comparisons of CD8+ T cells specific for human immunodeficiency virus, hepatitis C virus, and cytomegalovirus reveal differences in frequency, immunodominance, phenotype, and interleukin-2 responsiveness. J Virol 2009; 83:2728-42. [PMID: 19129459 PMCID: PMC2648273 DOI: 10.1128/jvi.02128-08] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2008] [Accepted: 12/24/2008] [Indexed: 11/20/2022] Open
Abstract
To better understand the components of an effective immune response to human immunodeficiency virus (HIV), the CD8(+) T-cell responses to HIV, hepatitis C virus (HCV), and cytomegalovirus (CMV) were compared with regard to frequency, immunodominance, phenotype, and interleukin-2 (IL-2) responsiveness. Responses were examined in rare patients exhibiting durable immune-mediated control over HIV, termed long-term nonprogressors (LTNP) or elite controllers, and patients with progressive HIV infection (progressors). The magnitude of the virus-specific CD8(+) T-cell response targeting HIV, CMV, and HCV was not significantly different between LTNP and progressors, even though their capacity to proliferate to HIV antigens was preserved only in LTNP. In contrast to HIV-specific CD8(+) T-cell responses of LTNP, HLA B5701-restricted responses within CMV pp65 were rare and did not dominate the total CMV-specific response. Virus-specific CD8(+) T cells were predominantly CD27(+)45RO(+) for HIV and CD27(-)45RA(+) for CMV; however, these phenotypes were highly variable and heavily influenced by the degree of viremia. Although IL-2 induced significant expansions of CMV-specific CD8(+) T cells in LTNP and progressors by increasing both the numbers of cells entering the proliferating pool and the number of divisions, the proliferative capacity of a significant proportion of HIV-specific CD8(+) T cells was not restored with exogenous IL-2. These results suggest that immunodominance by HLA B5701-restricted cells is specific to HIV infection in LTNP and is not a feature of responses to other chronic viral infections. They also suggest that poor responsiveness to IL-2 is a property of HIV-specific CD8(+) T cells of progressors that is not shared with responses to other viruses over which immunologic control is maintained.
Collapse
Affiliation(s)
- Prasanna Jagannathan
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892-1876, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Rodrigues L, Nandakumar S, Bonorino C, Rouse BT, Kumaraguru U. IL-21 and IL-15 cytokine DNA augments HSV specific effector and memory CD8+ T cell response. Mol Immunol 2009; 46:1494-504. [PMID: 19233474 DOI: 10.1016/j.molimm.2008.12.033] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Accepted: 12/24/2008] [Indexed: 02/02/2023]
Abstract
The recurrence of lesions and transmission of Herpes simplex virus is dependent on the number and function of viral specific CD8(+) T cells, especially the memory T cells. The generation, turnover and set point of this cell population is maintained by different factors like exposure to antigen, cytokines and co-stimulatory molecules. However, the contribution of these factors in the generation and maintenance of the memory CD8(+) T cell population is still controversial, since it is not clear if homeostatic proliferation driven by cytokines can overcome T cell receptor (TCR) signaling. Since, interleukin 15 (IL-15) and interleukin 21 (IL-21) are cytokines implicated in homeostatic control of CD8(+) T cell pool, we constructed and used expression plasmids coding for IL-15 (pIL-15) and IL-21 (pIL-21) to expand HSV specific CD8(+) T cells in an animal model. Our results showed that the IL-21 increased the frequency of CD8(+) T cells in the absence of antigen, although the magnitude of this response was dependent on TCR signaling. Both pIL-15 and pIL-21 boosted the numbers of antigen specific CD8(+) IFNgamma producing cells in the primary response. In the memory phase, numbers of CD8(+) CD44(high) as well as CD8(+) T cells producing IFN-gamma and TNF-alpha were increased when pIL-15 and pIL-21 were used alone or in combination, compared to vector treatment only, and association of antigen further increased the proliferative response. Our data suggest that genetic treatment with pIL-15 and pIL-21 in the presence or absence of cognate antigen can contribute to immune-enhancement against HSV.
Collapse
Affiliation(s)
- Luiz Rodrigues
- Faculdade de Biociências e Instituto de Pesquisas Biomédicas, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS 90619-900, Brazil
| | | | | | | | | |
Collapse
|